Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Elife ; 82019 07 10.
Article in English | MEDLINE | ID: mdl-31291191

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a condition characterized by absent puberty and infertility due to gonadotropin releasing hormone (GnRH) deficiency, which is often associated with anosmia (Kallmann syndrome, KS). We identified loss-of-function heterozygous mutations in anti-Müllerian hormone (AMH) and its receptor, AMHR2, in 3% of CHH probands using whole-exome sequencing. We showed that during embryonic development, AMH is expressed in migratory GnRH neurons in both mouse and human fetuses and unconvered a novel function of AMH as a pro-motility factor for GnRH neurons. Pathohistological analysis of Amhr2-deficient mice showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in reduced fertility in adults. Our findings highlight a novel role for AMH in the development and function of GnRH neurons and indicate that AMH signaling insufficiency contributes to the pathogenesis of CHH in humans.


Subject(s)
Anti-Mullerian Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypogonadism/metabolism , Neurons/metabolism , Signal Transduction , Adolescent , Adult , Amino Acid Sequence , Animals , Anti-Mullerian Hormone/genetics , Axons/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , COS Cells , Cell Movement , Chlorocebus aethiops , Female , Fertility , Fetus/metabolism , Heterozygote , Humans , Loss of Function Mutation , Luteinizing Hormone/metabolism , Male , Mice, Inbred C57BL , Olfactory Bulb/metabolism , Pedigree , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Young Adult
2.
Cell Mol Life Sci ; 75(22): 4223-4234, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29955905

ABSTRACT

The immune system plays a critical role in the establishment, development, and progression of head and neck squamous cell carcinoma (HNSCC). As treatment with single-immune checkpoint agent results in a lower response rate in patients, it is important to investigate new strategies to maintain favorable anti-tumor immune response. Herein, the combination immunotherapeutic value of CTLA4 blockade and SFKs inhibition was assessed in transgenic HNSCC mouse model. Our present work showed that tumor growth was not entirely controlled when HNSCC model mice were administered anti-CTLA4 chemotherapeutic treatment. Moreover, it was observed that Src family kinases (SFKs) were hyper-activated and lack of anti-tumor immune responses following anti-CTLA4 chemotherapeutic treatment. We hypothesized that activation of SFKs is a mechanism of anti-CTLA4 immunotherapy resistance. We, therefore, carried out combined drug therapy using anti-CTLA4 mAbs and an SFKs' inhibitor, dasatinib. As expected, dasatinib and anti-CTLA4 synergistically inhibited tumor growth in Tgfbr1/Pten 2cKO mice. Furthermore, dasatinib and anti-CTLA4 combined to reduce the number of myeloid-derived suppressor cells and Tregs, increasing the CD8+ T cell-to-Tregs ratio. We also found that combining dasatinib with anti-CTLA4 therapy significantly attenuated the expression of p-STAT3Y705 and Ki67 in tumoral environment. These results suggest that combination therapy with SFKs inhibitors may be a useful therapeutic approach to increase the efficacy of anti-CTLA4 immunotherapy in HNSCC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CTLA-4 Antigen/immunology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/therapy , Dasatinib/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Drug Therapy, Combination , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/therapy , Immunotherapy , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , STAT3 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , src-Family Kinases/antagonists & inhibitors
3.
Gastroenterology ; 153(5): 1378-1391.e6, 2017 11.
Article in English | MEDLINE | ID: mdl-28734833

ABSTRACT

BACKGROUND & AIMS: Transforming growth factor beta (TGF-ß) suppresses early stages of tumorigenesis, but also contributes to migration and metastasis of cancer cells. A large number of human tumors contain mutations that inactivate its receptors, or downstream proteins such as Smad transcription factors, indicating that the TGF-ß signaling pathway prevents tumor growth. We investigated the effects of TGF-ß inhibition on liver tumorigenesis in mice. METHODS: C57BL/6 mice received hydrodynamic tail-vein injections of transposons encoding HRASG12V and a short hairpin RNA (shRNA) to down-regulate p53, or those encoding HRASG12V and MYC, or those encoding HRASG12V and TAZS89A, to induce liver tumor formation; mice were also given injections of transposons encoding SMAD7 or shRNA against SMAD2, SMAD3, SMAD4, or SNAI1 (Snail), with or without ectopic expression of Snail. Survival times were compared, and livers were weighted and examined for tumors. Liver tumor tissues were analyzed by quantitative reverse-transcription PCR, RNA sequencing, immunoblots, and immunohistochemistry. We analyzed gene expression levels in human hepatocellular carcinoma samples deposited in The Cancer Genome Atlas. A cell proliferation assay was performed using human liver cancer cell lines (HepG2 and Huh7) stably expressing Snail or shRNA against Snail. RESULTS: TGF-ß inhibition via overexpression of SMAD7 (or knockdown of SMAD2, SMAD3, or SMAD4) consistently reduced formation and growth of liver tumors in mice that expressed activated RAS plus shRNA against p53, or in mice that expressed activated RAS and TAZ. TGF-ß signaling activated transcription of the Snail gene in liver tumors induced by HRASG12V and shRNA against p53, and by activated RAS and TAZ. Knockdown of Snail reduced liver tumor formation in both tumor models. Ectopic expression of Snail restored liver tumorigenesis suppressed by disruption of TGF-ß signaling. In human hepatocellular carcinoma, Snail expression correlated with TGF-ß activation. Ectopic expression of Snail increased cellular proliferation, whereas Snail knockdown led to reduced proliferation in human hepatocellular carcinoma cells. CONCLUSIONS: In analyses of transgenic mice, we found TGF-ß signaling to be required for formation of liver tumors upon expression of activated RAS and shRNA down-regulating p53, and upon expression of activated RAS and TAZ. Snail is the TGF-ß target that is required for hepatic tumorigenesis in these models.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms, Experimental/metabolism , Liver/metabolism , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , Genes, ras , Genetic Predisposition to Disease , Hep G2 Cells , Humans , Liver/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors/genetics , Time Factors , Transfection , Transforming Growth Factor beta/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Up-Regulation
4.
Arterioscler Thromb Vasc Biol ; 37(11): 2102-2113, 2017 11.
Article in English | MEDLINE | ID: mdl-28729364

ABSTRACT

OBJECTIVE: The role of TGF-ß (transforming growth factor-ß) signaling in abdominal aortic aneurysm (AAA) formation is controversial. Others reported that systemic blockade of TGF-ß by neutralizing antibodies accelerated AAA development in angiotensin II-infused mice. This result is consistent with other studies suggesting that TGF-ß signaling prevents AAA. Development of a therapy for AAA that exploits the protective actions of TGF-ß would be facilitated by identification of the mechanisms through which TGF-ß prevents AAA. We hypothesized that TGF-ß signaling prevents AAA by its actions on aortic medial smooth muscle cells. APPROACH AND RESULTS: We compared the prevalence, severity, and histopathology of angiotensin II-induced AAA among control mice (no TGF-ß blockade), mice with antibody-mediated systemic neutralization of TGF-ß, and mice with genetically based smooth muscle-specific loss of TGF-ß signaling. Surprisingly, we found that systemic-but not smooth muscle-specific-TGF-ß blockade significantly increased the prevalence of AAA and tended to increase AAA severity, adventitial thickening, and aortic wall macrophage accumulation. In contrast, abdominal aortas of mice with smooth muscle-specific loss of TGF-ß signaling differed from controls only in having a thinner media. We examined thoracic aortas of the same mice. Here we found that smooth muscle-specific loss of Tgfbr2-but not systemic TGF-ß neutralization-significantly accelerated development of aortic pathology, including increased prevalence of intramural hematomas, medial thinning, and adventitial thickening. CONCLUSION: Our results suggest that TGF-ß signaling prevents both abdominal and thoracic aneurysmal disease but does so by distinct mechanisms. Smooth muscle extrinsic signaling protects the abdominal aorta and smooth muscle intrinsic signaling protects the thoracic aorta.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal/prevention & control , Aortic Aneurysm, Thoracic/prevention & control , Muscle, Smooth, Vascular/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Adventitia/metabolism , Adventitia/pathology , Animals , Antibodies/pharmacology , Aorta, Abdominal/drug effects , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Thoracic/chemically induced , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Dilatation, Pathologic , Disease Models, Animal , Female , Genetic Predisposition to Disease , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Severity of Illness Index , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/antagonists & inhibitors , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/antagonists & inhibitors , Transforming Growth Factor beta3/metabolism , Tunica Media/metabolism , Tunica Media/pathology , Vascular Remodeling/drug effects
5.
J Pathol ; 243(1): 89-99, 2017 09.
Article in English | MEDLINE | ID: mdl-28657664

ABSTRACT

Although a putative role for transforming growth factor-ß (TGFB) signalling in the pathogenesis of human endometrial cancer has long been proposed, the precise function of TGFB signalling in the development and progression of endometrial cancer remains elusive. Depletion of phosphatase and tensin homologue (PTEN) in the mouse uterus causes endometrial cancer. To identify the potential role of TGFB signalling in endometrial cancer, we simultaneously deleted TGFB receptor 1 (Tgfbr1) and Pten in the mouse uterus by using Cre-recombinase driven by the progesterone receptor (termed Ptend/d ;Tgfbr1d/d ). We found that Ptend/d ;Tgfbr1d/d mice developed severe endometrial lesions that progressed more rapidly than those resulting from conditional deletion of Pten alone, suggesting that TGFB signalling synergizes with PTEN to suppress endometrial cancer progression. Remarkably, Ptend/d ;Tgfbr1d/d mice developed distant pulmonary metastases, leading to a significantly reduced lifespan. The development of metastasis and accelerated tumour progression in Ptend/d ;Tgfbr1d/d mice are associated with increased production of proinflammatory chemokines, enhanced cancer cell motility, as shown by myometrial invasion and disruption, and an altered tumour microenvironment characterized by recruitment of tumour-associated macrophages. Thus, conditional deletion of Tgfbr1 in PTEN-inactivated endometrium leads to a disease that recapitulates invasive and lethal human endometrial cancer. This mouse model may be valuable for preclinical testing of new cancer therapies, particularly those targeting metastasis, one of the hallmarks of cancer and a major cause of death in endometrial cancer patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Endometrial Neoplasms/enzymology , Endometrium/enzymology , PTEN Phosphohydrolase/deficiency , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Animals , Cell Movement , Cell Proliferation , Chemokines/metabolism , Disease Progression , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Gene Deletion , Genetic Predisposition to Disease , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Macrophages/metabolism , Macrophages/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , Phenotype , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Time Factors , Tumor Burden , Tumor Microenvironment
6.
PLoS One ; 12(5): e0177826, 2017.
Article in English | MEDLINE | ID: mdl-28542404

ABSTRACT

Recent emerging data indicate that the increase in the expression and activity of the transforming growth factor beta 1 (Tgf-ß1) signaling may have detrimental effect to mature articular cartilage of knee joints. However, there is no information about whether or not this is the case in condylar cartilages. The objective of this study is to investigate the protein expression and activity of Tgf-ß1 signaling in degenerative condylar cartilages. We also investigate biological effects of the conditional deletion of transforming growth factor receptor type II (Tgfbr2) in condylar cartilage of adult mice after a partial discectomy. Two mouse models of osteoarthritis (OA) were used to examine protein expressions of Tgf-ß1 and p-Smad2/3 in condylar cartilages at early degenerative stages. In addition, cartilage specific Tgfbr2-deficient adult mice were subjected to a partial discectomy. The morphological condition of condylar cartilages was evaluated in mice at 4 and 12 weeks after the surgery. We found that protein levels of Tgf-ß1 and p-Smad2/3 were increased in the degenerative condylar cartilage of the mouse models. The conditional removal of Tgfbr2 in mature condylar cartilage significantly delayed the progressive progression of the cartilage degeneration induced by a partial discectomy. We conclude that the increase in the expression and activity of Tgf-ß1 signaling may have detrimental effect to mature condylar cartilages. Therefore, inhibition of Tgf-ß1 signaling may be able to protect condylar cartilages from being degraded in mature temporomandibular joints.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/surgery , Signal Transduction , Transforming Growth Factor beta1/metabolism , Aggrecans/metabolism , Animals , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/deficiency , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Smad2 Protein/metabolism , Smad3 Protein/metabolism
7.
Int J Mol Sci ; 18(4)2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28420207

ABSTRACT

The transforming growth factor-ß (TGFß) family signaling pathways play an important role in regulatory cellular networks and exert specific effects on developmental programs during embryo development. However, the function of TGFß signaling pathways on the early kidney development remains unclear. In this work, we aim to detect the underlying role of TGFß type II receptor (TßRII) in vitro, which has a similar expression pattern as the crucial regulator Six2 during early kidney development. Firstly, the 5-ethynyl-2'-deoxyuridine (EdU) assay showed knock down of TßRII significantly decreased the proliferation ratio of metanephric mesenchyme (MM) cells. Additionally, real-time Polymerase Chain Reaction (PCR) and Western blot together with immunofluorescence determined that the mRNA and protein levels of Six2 declined after TßRII knock down. Also, Six2 was observed to be able to partially rescue the proliferation phenotype caused by the depletion of TßRII. Moreover, bioinformatics analysis and luciferase assay indicated Smad3 could transcriptionally target Six2. Further, the EdU assay showed that Smad3 could also rescue the inhibition of proliferation caused by the knock down of TßRII. Taken together, these findings delineate the important function of the TGFß signaling pathway in the early development of kidney and TßRII was shown to be able to promote the expression of Six2 through Smad3 mediating transcriptional regulation and in turn activate the proliferation of MM cells.


Subject(s)
Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Cell Proliferation , Gene Expression , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Kidney/embryology , Kidney/metabolism , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
8.
Elife ; 62017 02 21.
Article in English | MEDLINE | ID: mdl-28219480

ABSTRACT

Squamous cell carcinomas occurring at transition zones are highly malignant tumors with poor prognosis. The identity of the cell population and the signaling pathways involved in the progression of transition zone squamous cell carcinoma are poorly understood, hence representing limited options for targeted therapies. Here, we identify a highly tumorigenic cancer stem cell population in a mouse model of transitional epithelial carcinoma and uncover a novel mechanism by which loss of TGFß receptor II (Tgfbr2) mediates invasion and metastasis through de-repression of ELMO1, a RAC-activating guanine exchange factor, specifically in cancer stem cells of transition zone tumors. We identify ELMO1 as a novel target of TGFß signaling and show that restoration of Tgfbr2 results in a complete block of ELMO1 in vivo. Knocking down Elmo1 impairs metastasis of carcinoma cells to the lung, thereby providing insights into the mechanisms of progression of Tgfbr2-deficient invasive transition zone squamous cell carcinoma.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/physiopathology , Cell Movement , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/physiology , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Animals , Disease Models, Animal , Mice , Neoplasm Metastasis , Receptor, Transforming Growth Factor-beta Type II
9.
Anticancer Res ; 37(2): 499-513, 2017 02.
Article in English | MEDLINE | ID: mdl-28179295

ABSTRACT

Intercellular reactive oxygen/reactive nitrogen species-(ROS/RNS)-dependent induction of apoptosis in malignant cells is discussed as a potential control step during oncogenesis. In previous studies, the mechanism of intercellular apoptosis-inducing signaling was mainly established through the use of specific inhibitors and scavengers. Here, a detailed analysis was carried out based on small interfering ribonucleic acid (siRNA)-mediated knockdown of central players of intercellular ROS/RNS signaling and of the mitochondrial and the FAS receptor-dependent pathway of apoptosis. The data show that transforming growth factor ß1, transforming growth factor ß receptor, NADPH oxidase-1 (NOX1), NOX1 organizer, and NOX1 activator control the HOCl and the NO/peroxynitrite signaling pathways. Dual oxidase-1 (DUOX1) is specifically involved in HOCl signaling, and NO synthase in NO/peroxynitrite signaling. Both pathways utilize intracellular signal transduction through protein kinase C zeta, sphingomyelinase and central elements of the mitochondrial pathway of apoptosis, whereas the FAS receptor and FAS ligand do not seem to play a role.


Subject(s)
Apoptosis/physiology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Cell Line, Transformed , Cell Line, Tumor , Gene Knockdown Techniques , Humans , NADH, NADPH Oxidoreductases/deficiency , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rats , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
10.
Int J Cancer ; 140(4): 853-863, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27790711

ABSTRACT

Colorectal cancer (CRC) results from the accumulation of gene mutations and epigenetic alterations in colon epithelial cells, which promotes CRC formation through deregulating signaling pathways. One of the most commonly deregulated signaling pathways in CRC is the transforming growth factor ß (TGF-ß) pathway. Importantly, the effects of TGF-ß signaling inactivation in CRC are modified by concurrent mutations in the tumor cell, and these concurrent mutations determine the ultimate biological effects of impaired TGF-ß signaling in the tumor. However, many of the mutations that cooperate with the deregulated TGF-ß signaling pathway in CRC remain unknown. Therefore, we sought to identify candidate driver genes that promote the formation of CRC in the setting of TGF-ß signaling inactivation. We performed a forward genetic screen in mice carrying conditionally inactivated alleles of the TGF-ß receptor, type II (Tgfbr2) using Sleeping Beauty (SB) transposon mediated mutagenesis. We used TAPDANCE and Gene-centric statistical methods to identify common insertion sites (CIS) and, thus, candidate tumor suppressor genes and oncogenes within the tumor genome. CIS analysis of multiple neoplasms from these mice identified many candidate Tgfbr2 cooperating genes and the Wnt/ß-catenin, Hippo and MAPK pathways as the most commonly affected pathways. Importantly, the majority of candidate genes were also found to be mutated in human CRC. The SB transposon system provides an unbiased method to identify Tgfbr2 cooperating genes in mouse CRC that are functionally relevant and that may provide further insight into the pathogenesis of human CRC.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Colorectal Neoplasms/genetics , DNA Transposable Elements , Genes, Neoplasm , Genes, Tumor Suppressor , Genetic Association Studies/methods , Mutagenesis, Insertional , Neoplasm Proteins/physiology , Signal Transduction/genetics , Transforming Growth Factor beta/physiology , Adenocarcinoma/metabolism , Adenoma/metabolism , Animals , Colorectal Neoplasms/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Sequence Analysis, DNA , Signal Transduction/physiology , Species Specificity
11.
Int J Cancer ; 140(5): 1173-1185, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27798955

ABSTRACT

SRC family kinases (SFKs), a group of nonreceptor tyrosine kinases, modulate multiple cellular functions, such as cell proliferation, differentiation and metabolism. SFKs display aberrant activity in progressive stages of human cancers. However, the precise role of SFKs in the head and neck squamous cell carcinoma (HNSCC) signaling network is far from clear. In this study, we found that the inhibition of SFKs activity by dasatinib effectively reduced the tumor size and population of MDSCs in the HNSCC mouse model. Molecular analysis indicates that phosphorylation of LYN, rather than SRC, was inhibited by dasatinib treatment. Next, we analyzed LYN expression by immunostaining and found that it was overexpressed in the human HNSCC specimens. Moreover, LYN expression in stromal cells positively correlated with myeloid-derived suppressor cells (MDSCs) makers CD11b and CD33 in human HNSCC. The dual positive expression of LYN in epithelial and stromal cells (EPI+ SRT+ ) was associated with unfavorable overall survival of HNSCC patients. These findings indicate that SFKs may be a potential target for an effective immunotherapy of HNSCC by decreasing MDSCs and moreover, LYN will have an impact on such therapeutic strategy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Dasatinib/therapeutic use , Head and Neck Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/immunology , Dasatinib/pharmacology , Enzyme Induction/drug effects , Epithelial Cells/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/immunology , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/deficiency , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/deficiency , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Stromal Cells/enzymology , Tumor Escape , Up-Regulation , Xenograft Model Antitumor Assays , src-Family Kinases/biosynthesis , src-Family Kinases/genetics , src-Family Kinases/metabolism , src-Family Kinases/physiology
12.
PLoS One ; 11(11): e0166422, 2016.
Article in English | MEDLINE | ID: mdl-27835699

ABSTRACT

Mutations in TGFBR2, a component of the transforming growth factor (TGF)-ß signaling pathway, occur in high-frequency microsatellite instability (MSI-H) colorectal cancer (CRC). In mouse models, Tgfbr2 inactivation in the intestinal epithelium accelerates the development of malignant intestinal tumors in combination with disruption of the Wnt-ß-catenin pathway. However, no studies have further identified the genes influenced by TGFBR2 inactivation following disruption of the Wnt-ß-catenin pathway. We previously described CDX2P-G19Cre;Apcflox/flox mice, which is stochastically null for Apc in the colon epithelium. In this study, we generated CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice, with simultaneous loss of Apc and Tgfbr2. These mice developed tumors, including adenocarcinoma in the proximal colon. We compared gene expression profiles between tumors of the two types of mice using microarray analysis. Our results showed that the expression of the murine homolog of GSDMC was significantly upregulated by 9.25-fold in tumors of CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice compared with those of CDX2P-G19Cre;Apcflox/flox mice. We then investigated the role of GSDMC in regulating CRC tumorigenesis. The silencing of GSDMC led to a significant reduction in the proliferation and tumorigenesis of CRC cell lines, whereas the overexpression of GSDMC enhanced cell proliferation. These results suggested that GSDMC functioned as an oncogene, promoting cell proliferation in colorectal carcinogenesis. In conclusion, combined inactivation of both Apc and Tgfbr2 in the colon epithelium of a CRC mouse model promoted development of adenocarcinoma in the proximal colon. Moreover, GSDMC was upregulated by TGFBR2 mutation in CRC and promoted tumor cell proliferation in CRC carcinogenesis, suggesting that GSDMC may be a promising therapeutic target.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/metabolism , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Microarray Analysis , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/deficiency , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
Sci Rep ; 6: 35444, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739498

ABSTRACT

Transforming growth factor (TGF)-ß signaling disorder has emerged as a common molecular signature for aortic aneurysm development. The timing of postnatal maturation plays a key role in dictating the biological outcome of TGF-ß signaling disorders in the aortic wall. In this study, we investigated the impact of deficiency of TGFß receptors on the structural homeostasis of mature aortas. We used an inducible Cre-loxP system driven by a Myh11 promoter to delete Tgfbr1, Tgfbr2, or both in smooth muscle cells (SMCs) of adult mice. TGFBR1 deficiency resulted in rapid and severe aneurysmal degeneration, with 100% penetrance of ascending thoracic aortas, whereas TGFBR2 deletion only caused mild aortic pathology with low (26%) lesion prevalence. Removal of TGFBR2 attenuated the aortic pathology caused by TGFBR1 deletion and correlated with a reduction of early ERK phosphorylation. In addition, the production of angiotensin (Ang)-converting enzyme was upregulated in TGFBR1 deficient aortas at the early stage of aneurysmal degeneration. Inhibition of ERK phosphorylation or blockade of AngII type I receptor AT1R prevented aneurysmal degeneration of TGFBR1 deficient aortas. In conclusion, loss of SMC-Tgfbr1 triggers multiple deleterious pathways, including abnormal TGFBR2, ERK, and AngII/AT1R signals that disrupt aortic wall homeostasis to cause aortic aneurysm formation.


Subject(s)
Aortic Aneurysm/genetics , MAP Kinase Signaling System , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Angiotensin II/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/metabolism , Peptidyl-Dipeptidase A/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Angiotensin/metabolism , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism
14.
PLoS One ; 11(8): e0159710, 2016.
Article in English | MEDLINE | ID: mdl-27505173

ABSTRACT

The epicardium plays an important role in coronary vessel formation and Tgfbr3-/- mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3-/- epicardial cells display the same defects. Tgfbr3+/+ and Tgfbr3-/- cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFßR3 (TGFß1, TGFß2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3+/+ and Tgfbr3-/- cells when incubated with VEH (604), TGFß1 (515), TGFß2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3-/- cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFß1-TGFß2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3+/+ and Tgfbr3-/- cells after TGFß and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3-/- cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3+/+ epicardial cells after TGFß2 or BMP2 incubation, while Tgfbr3-/- cells failed to activate NF-ĸB in response to these ligands. Tgfbr3+/+ epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFß2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3-/- epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFßR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFßR3 signaling.


Subject(s)
Pericardium/cytology , Pericardium/embryology , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transcriptome , Animals , Cell Line , Ligands , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Proteoglycans/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Sequence Analysis, RNA
15.
Eur J Pharmacol ; 789: 134-143, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27397431

ABSTRACT

Renal fibrosis, a common feature and leading cause for End Stage Renal Disease, still lacks effective therapy. In the current study, we detected and compared the anti-fibrotic effects of wogonin and wogonoside, two major components of Scutellaria baicalensis Georgi, in TGF-ß1-treated tubular epithelial cells of human and murine origins. Results consistently showed that compared with wogonoside, wogonin inhibits TGF-ß1-induced upregulated mRNA and protein levels of collagen I and α-SMA with more efficiency, which was further confirmed by the immunofluorescence results that wogonin decreased the percentage of collagen I and α-SMA positive cells in TGF-ß1-treated tubular epithelial cells. Mechanistically, wogonin mainly decreased Smad3 phosphorylation, but had marginal effect on non-canonical TGF-ß signaling pathways, such as p38 and ERK MAP Kinase. Furthermore, in the cells deficient for TGF-ß signaling or downstream Smad3, results demonstrated that even high concentration of wogonin failed to further decrease the level of collagen I and α-SMA, indicating the essential role of TGF-ß/Smad3 signaling inhibition in the therapeutic action of wogonin in TGF-ß1-stimulated tubular epithelial cells. Collectively, our results indicated that wogonin may be utilized as a potential anti-fibrotic Traditional Chinese Medicine monomer in the treatment of renal fibrosis.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/pathology , Flavanones/pharmacology , Glucosides/pharmacology , Kidney Tubules, Proximal/pathology , Smad3 Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Fibrosis , Flavanones/therapeutic use , Glucosides/therapeutic use , Humans , Mice , Phosphorylation/drug effects , Rats , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism , Smad3 Protein/deficiency , Transforming Growth Factor beta1/pharmacology
16.
Clin Exp Immunol ; 185(2): 154-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27148790

ABSTRACT

During chronic inflammation, interleukin (IL)-22 expression is up-regulated in both CD4 and CD8 T cells, exerting a protective role in infections. However, in autoimmunity, IL-22 appears to have either a protective or a pathogenic role in a variety of murine models of autoimmunity and, by extrapolation, in humans. It is not clear whether IL-22 itself mediates inflammation or is a by-product of inflammation. We have taken advantage of the dominant negative form of transforming growth factor beta receptor type II (dnTGF-ßRII) mice that develop both inflammatory bowel disease and autoimmune cholangitis and studied the role and the biological function of IL-22 by generating IL-22(-/-) dnTGF-ßRII mice. Our data suggest that the influence of IL-22 on autoimmunity is determined in part by the local microenvironment. In particular, IL-22 deficiency exacerbates tissue injury in inflammatory bowel disease, but has no influence on either the hepatocytes or cholangiocytes in the same model. These data take on particular significance in the previously defined effects of IL-17A, IL-12p40 and IL-23p19 deficiency and emphasize that, in colitis, there is a dominant role of IL-23/T helper type 17 (Th17) signalling. Furthermore, the levels of IL-22 are IL-23-dependent. The use of cytokine therapy in patients with autoimmune disease has significant potential, but must take into account the overlapping and often promiscuous effects that can theoretically exacerbate inflammation.


Subject(s)
Autoimmune Diseases/immunology , Cholangitis/immunology , Inflammatory Bowel Diseases/prevention & control , Interleukins/metabolism , Receptors, Transforming Growth Factor beta/deficiency , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammatory Bowel Diseases/immunology , Interleukin-17/analysis , Interleukin-23/metabolism , Interleukins/deficiency , Interleukins/immunology , Liver Cirrhosis, Biliary/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transforming Growth Factor beta/genetics , Interleukin-22
17.
Arterioscler Thromb Vasc Biol ; 36(5): 919-27, 2016 05.
Article in English | MEDLINE | ID: mdl-26988590

ABSTRACT

OBJECTIVE: Transforming growth factor-beta is a pleiotropic cytokine having diverse roles in vascular morphogenesis, homeostasis, and pathogenesis. Altered activity of and signaling through transforming growth factor-beta has been implicated in thoracic aortic aneurysms and dissections, conditions characterized by a reduced structural integrity of the wall that associates with altered biomechanics and mechanobiology. We quantify and contrast the passive and active biaxial biomechanical properties of the ascending and proximal descending thoracic aorta in a mouse model of altered transforming growth factor-beta signaling, with and without treatment with rapamycin. APPROACH AND RESULTS: Postnatal disruption of the gene (Tgfbr2) that codes the type II transforming growth factor-beta receptor compromises vessel-level contractility and elasticity. Daily treatment with rapamycin, a mechanistic target of rapamycin inhibitor that protects against aortic dissection in these mice, largely preserves or restores the contractile function while the passive properties remain compromised. Importantly, this increased smooth muscle contractility protects an otherwise vulnerable aortic wall from pressure-induced intramural delaminations in vitro. CONCLUSIONS: Notwithstanding the protection afforded by rapamycin in vivo and in vitro, the residual mechanical dysfunctionality suggests a need for caution if rapamycin is to be considered as a potential therapeutic. There is a need for in vivo evaluations in cases of increased hemodynamic loading, including hypertension or extreme exercise, which could unduly stress a structurally vulnerable aortic wall. Given these promising early results, however, such studies are clearly warranted.


Subject(s)
Aortic Aneurysm, Thoracic/prevention & control , Aortic Dissection/prevention & control , Extracellular Matrix/drug effects , Muscle, Smooth, Vascular/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Sirolimus/pharmacology , Vascular Stiffness/drug effects , Vasoconstriction/drug effects , Aortic Dissection/genetics , Aortic Dissection/metabolism , Aortic Dissection/physiopathology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/physiopathology , Arterial Pressure , Elasticity , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Genetic Predisposition to Disease , Male , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Phenotype , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Stress, Mechanical , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
18.
Proc Natl Acad Sci U S A ; 113(8): 2294-9, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26842836

ABSTRACT

Rupture of the ovarian follicle releases the oocyte at ovulation, a timed event that is critical for fertilization. It is not understood how the protease activity required for rupture is directed with precise timing and localization to the outer surface, or apex, of the follicle. We hypothesized that vasoconstriction at the apex is essential for rupture. The diameter and blood flow of individual vessels and the thickness of the apical follicle wall were examined over time to expected ovulation using intravital multiphoton microscopy. Vasoconstriction of apical vessels occurred within hours preceding follicle rupture in wild-type mice, but vasoconstriction and rupture were absent in Amhr2(cre/+)SmoM2 mice in which follicle vessels lack the normal association with vascular smooth muscle. Vasoconstriction is not simply a response to reduced thickness of the follicle wall; vasoconstriction persisted in wild-type mice when thinning of the follicle wall was prevented by infusion of protease inhibitors into the ovarian bursa. Ovulation was inhibited by preventing the periovulatory rise in the expression of the vasoconstrictor endothelin 2 by follicle cells of wild-type mice. In these mice, infusion of vasoconstrictors (either endothelin 2 or angiotensin 2) into the bursa restored the vasoconstriction of apical vessels and ovulation. Additionally, infusion of endothelin receptor antagonists into the bursa of wild-type mice prevented vasoconstriction and follicle rupture. Processing tissue to allow imaging at increased depth through the follicle and transabdominal ultrasonography in vivo showed that decreased blood flow is restricted to the apex. These results demonstrate that vasoconstriction at the apex of the follicle is essential for ovulation.


Subject(s)
Ovarian Follicle/blood supply , Ovarian Follicle/physiology , Ovulation/physiology , Vasoconstriction/physiology , Animals , Endothelin-2/deficiency , Endothelin-2/genetics , Endothelin-2/physiology , Female , Intravital Microscopy , Mice , Mice, 129 Strain , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Ovarian Follicle/diagnostic imaging , Ovulation/genetics , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Receptors, Peptide/physiology , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Smoothened Receptor , Ultrasonography , Vasoconstriction/genetics
19.
Am J Physiol Renal Physiol ; 310(6): F499-510, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26739889

ABSTRACT

Transforming growth factor-ß (TGF-ß) and hepatocyte growth factor (HGF) play key roles in regulating the response to renal injury but are thought to mediate divergent effects on cell behavior. However, how TGF-ß signaling alters the response to HGF in epithelia, the key site of HGF signaling in the injured kidney, is not well studied. Contrary to our expectation, we showed that deletion of the TGF-ß type II receptor in conditionally immortalized proximal tubule (PT) cells impaired HGF-dependent signaling. This reduced signaling was due to decreased transcription of c-Met, the HGF receptor, and the TGF-ß-dependent c-Met transcription and increased response to HGF in PT cells were mediated by the Notch pathway. The interactions of TGF-ß, HGF, and Notch pathways had biologically significant effects on branching morphogenesis, cell morphology, migration, and proliferation. In conclusion, epithelial TGF-ß signaling promotes HGF signaling in a Notch-dependent pathway. These findings suggest that TGF-ß modulates PT responses not only by direct effects, but also by affecting other growth factor signaling pathways.


Subject(s)
Hepatocyte Growth Factor/metabolism , Kidney Tubules, Proximal/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cells, Cultured , Mice , Protein Serine-Threonine Kinases/deficiency , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
20.
Exp Dermatol ; 25(2): 85-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26476131

ABSTRACT

Keratoacanthoma (KA) are common but exceptional benign tumors, often appearing on sun-exposed areas of light skinned people and showing spontaneous resolution. The goal of this study was to review existing literature, to point out the etiological complexity of KA biology and to answer the controversial debate if or not KA is a distinct entity or a variant of squamous cell carcinoma (SCC). Relying on recent results, we highlight that KA is an individual lesion with a unique molecular signature caused by alterations in the TGFß signalling pathway. These recent findings will help to understand the nature of KA and to develop new reliable diagnostic tools, simplifying the discrimination of the histologically similar KA and SCC.


Subject(s)
Keratoacanthoma , Skin Diseases , Carcinoma, Squamous Cell/diagnosis , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Comparative Genomic Hybridization , Diagnosis, Differential , Disease Progression , Genetic Predisposition to Disease , Humans , Keratoacanthoma/diagnosis , Keratoacanthoma/etiology , Keratoacanthoma/genetics , Keratoacanthoma/metabolism , Keratoacanthoma/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Radiation-Induced/chemistry , Neoplasms, Radiation-Induced/diagnosis , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction , Skin Diseases/diagnosis , Skin Diseases/etiology , Skin Diseases/genetics , Skin Diseases/metabolism , Skin Diseases/pathology , Skin Neoplasms/diagnosis , Sunlight/adverse effects , Transforming Growth Factor beta/physiology , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...